Immunity
Volume 55, Issue 6, 14 June 2022, Pages 982-997.e8
Journal home page for Immunity

Article
Dendritic cells can prime anti-tumor CD8+ T cell responses through major histocompatibility complex cross-dressing

https://doi.org/10.1016/j.immuni.2022.04.016Get rights and content

Highlights

  • Optimal anti-tumor CD8+ T cell priming requires MHC-I expression on cancer cells

  • Tumor-resident DCs and macrophages dress with cancer cell MHC-I in vivo

  • MHC-I cross-dressing is sufficient for antigen-specific CD8+ T cell priming ex vivo

  • The impact of MHC-I cross-dressing on CD8+ T cell priming differs between tumor models

Summary

Antigen cross-presentation, wherein dendritic cells (DCs) present exogenous antigen on major histocompatibility class I (MHC-I) molecules, is considered the primary mechanism by which DCs initiate tumor-specific CD8+ T cell responses. Here, we demonstrate that MHC-I cross-dressing, an antigen presentation pathway in which DCs acquire and display intact tumor-derived peptide:MHC-I molecules, is also important in orchestrating anti-tumor immunity. Cancer cell MHC-I expression was required for optimal CD8+ T cell activation in two subcutaneous tumor models. In vivo acquisition of tumor-derived peptide:MHC-I molecules by DCs was sufficient to induce antigen-specific CD8+ T cell priming. Transfer of tumor-derived human leukocyte antigen (HLA) molecules to myeloid cells was detected in vitro and in human tumor xenografts. In conclusion, MHC-I cross-dressing is crucial for anti-tumor CD8+ T cell priming by DCs. In addition to quantitatively enhancing tumor antigen presentation, MHC cross-dressing might also enable DCs to more faithfully and efficiently mirror the cancer cell peptidome.

Introduction

The anti-tumor immune response has been intensely studied over the past several decades (Vesely et al., 2011), leading to the development and widespread administration of cancer immunotherapies in the clinic (Baumeister et al., 2016; Waldman et al., 2020) However, cancer immunotherapy is not universally effective (Dougan et al., 2019), and progress is hindered by persisting gaps in our understanding of the fundamental mechanisms required for the orchestration of effective anti-cancer immune responses, particularly at the level of early T cell priming. Although the mechanisms by which CD8+ T cells are primed against human cancers remain unclear, murine models have provided critical insights into these questions. Recently, the professional antigen presenting cells (APCs) involved in anti-tumor T cell priming have been identified (Roberts et al., 2016; Salmon et al., 2016); however, the specific pathways utilized by these APCs to acquire and present tumor antigens in vivo remain incompletely defined.

Migratory CD103+ and CD11b+ dendritic cells (DCs) exist in tissues throughout the body, constitutively acquiring proteins from surrounding cells before trafficking to draining lymph nodes, where they present derivative peptide antigens to T cells in the context of major histocompatibility complex (MHC) molecules (Merad et al., 2013; Worbs et al., 2017). Migratory DCs within tumors similarly acquire and subsequently present cancer cell-derived antigens to T cells in tumor-draining lymph nodes (tdLNs) (Wculek et al., 2020). Recent evidence indicates that tumor antigens are exclusively transported to tdLNs by migratory CD103+ DCs (Ruhland et al., 2020; Salmon et al., 2016) and that these DCs are primarily responsible for anti-tumor CD8+ T cell priming in vivo (Roberts et al., 2016; Salmon et al., 2016). Migratory CD103+ DCs, along with lymph node resident CD8α+ DCs, comprise the BATF3- and IRF8-dependent conventional type 1 DC (cDC1) lineage (Anderson et al., 2021; Edelson et al., 2010; Hildner et al., 2008). Because antigen cross-presentation is a canonical function of cDC1 (Dudziak et al., 2007; Hildner et al., 2008) and because numerous studies have demonstrated a requirement for cDC1 in the activation of anti-tumor CD8+ T cell responses (Fuertes et al., 2011; Hildner et al., 2008; Wculek et al., 2020), prevailing thought is that tumor-specific CD8+ T cells are primed exclusively through antigen cross-presentation (Lee et al., 2020; Sánchez-Paulete et al., 2017).

However, an alternative antigen presentation mechanism has been described, in which DCs acquire and present intact peptide:MHC (pMHC) captured directly from neighboring cells (André et al., 2004; Dolan et al., 2006; Herrera et al., 2004; Russo et al., 2000; Wolfers et al., 2001). This phenomenon, known as MHC cross-dressing, has been implicated in antigen presentation in various contexts ranging from viral infection (Wakim and Bevan, 2011) and vaccination (Li et al., 2012) to thymic selection (Koble and Kyewski, 2009; Kroger et al., 2017; Perry et al., 2018), graft rejection (Liu et al., 2016), and peripheral tolerance to maternal microchimerism (Bracamonte-Baran et al., 2017). By necessity, conclusions regarding the role of MHC cross-dressing in antigen presentation by DCs have largely arisen from in vitro studies and in vivo models of MHC-mismatched bone marrow chimeric mice (Dolan et al., 2006; Koble and Kyewski, 2009; Wakim and Bevan, 2011) or solid organ transplantation (Liu et al., 2016), due to difficulty in controlling for cross-presentation without entirely ablating antigen presentation on MHC-I. Indeed, although MHC cross-dressing by DCs is sufficient to induce T cell priming in various contexts, the necessity of this antigen presentation pathway in mediating in vivo T cell activation has never been conclusively demonstrated in syngeneic hosts.

In this study, we employed two syngeneic murine tumor models expressing distinct model antigens presented in the context of the MHC-I molecule, H-2Kb (Kb), to determine the extent to which MHC-I cross-dressing was involved in antigen-specific CD8+ T cell priming. Although the magnitude of the effect varied, CD8+ T cell priming against Kb-restricted tumor antigens was impaired in mice harboring Kb−/− tumors, despite the cross-presentation pathway being fully intact. Furthermore, cancer cell-derived MHC-I molecules were readily observed within and on the surface of tumor-resident APCs, and MHC-I-deficient CD103+ cDC1 isolated from tdLNs of mice bearing Kb-sufficient tumors stimulated antigen-specific CD8+ T cells ex vivo. The importance of WDFY4-dependent antigen cross-presentation (Theisen et al., 2018) in mediating anti-tumor CD8+ T cell responses differed across experimental models, but in some cases, it was dispensable for in vivo CD8+ T cell priming. Finally, we observed that APCs became cross-dressed with human leukocyte antigen class I (HLA-I) molecules upon co-culture with HLA-mismatched tumor cells and in tumors xenografted in immunodeficient mice. Acquisition of tumor cell-derived HLA-I molecules correlated with uptake of tumor antigens in vitro. Similarly, APCs isolated from murine tumors acquired both tumor-derived MHC-I and fluorescent antigen in vivo, suggesting that MHC-I cross-dressing and internalization of tumor material may be linked processes. Taken together, our results demonstrate that MHC-I cross-dressing is central to the ability of cDC1 to orchestrate anti-tumor CD8+ T cell responses.

Section snippets

Loss of H-2Kb does not impact model antigen expression in cancer cells

In order to define the impact of cancer cell MHC-I expression on anti-tumor CD8+ T cell priming, two tumor models were utilized, C1498 leukemia and B16.F10 melanoma (both H-2b). These models were selected due to differences in cell of origin and baseline MHC-I expression, and both have been extensively characterized, with well-established growth kinetics in syngeneic C57BL/6 mice (LaBelle et al., 2002; Overwijk and Restifo, 2000). Moreover, the timing and magnitude of endogenous

Discussion

The finding that MHC-I cross-dressing by migratory CD103+ cDC1 is a critical antigen presentation pathway for mounting anti-tumor CD8+ T cell responses challenges our current understanding of the mechanisms by which DCs present cancer antigens. Our observations also raise the following question: Why does MHC-I cross-dressing appear to play such an important role in tumor antigen presentation when cross-presentation is a hallmark cDC1 function? Any definitive answer will require additional

Key resources table

Reagent or ResourceSourceIdentifier
Antibodies
CD16/32 - unconjugated (clone: 2.4g2)University of Chicago Cytometry and Antibody Technology FacilityN/A
CD3 - Biotin (clone: 17A2)BiolegendCat# 100244
CD3 - FITC (clone: 17A2)BiolegendCat# 100204
CD4 - Biotin (clone: GK1.5)BiolegendCat# 100404
CD4 - PE/Cy7 (clone: GK1.5)BiolegendCat# 100422
CD4 - APC (clone: GK1.5)BiolegendCat# 100412
CD8a - Biotin (clone: 53-6.7)BiolegendCat# 100704
CD8a - Pacific Blue (clone: 53-6.7)BiolegendCat# 100725
CD8a - BV605

Acknowledgments

We would like to thank Drs. T. Gajewski, A. Bendelac, T. Schumacher, and S. Springer for sharing mice, cell lines, and constructs central to this research. Special thanks to L. Degenstein of the University of Chicago Transgenics/ES Cell Technology Mouse Core Facility for help with microinjections. Thanks also to Dr. P. Savage for careful review of the manuscript and insightful conversations during the project. The authors have no financial interests to disclose. This work was funded by R01

References (61)

  • A. Savina et al.

    NOX2 controls phagosomal pH to regulate antigen processing during crosspresentation by dendritic cells

    Cell

    (2006)
  • A. Savina et al.

    The small GTPase Rac2 controls phagosomal alkalinization and antigen crosspresentation selectively in CD8(+) dendritic cells

    Immunity

    (2009)
  • M.T. Spiotto et al.

    Increasing tumor antigen expression overcomes “ignorance” to solid tumors via crosspresentation by bone marrow-derived stromal cells

    Immunity

    (2002)
  • J.-G. Zhang et al.

    The dendritic cell receptor Clec9A binds damaged cells via exposed actin filaments

    Immunity

    (2012)
  • D.A. Anderson et al.

    Genetic models of human and mouse dendritic cell development and function

    Nat. Rev. Immunol.

    (2021)
  • F. André et al.

    Exosomes as potent cell-free peptide-based vaccine. I. Dendritic cell-derived exosomes transfer functional MHC class I/peptide complexes to dendritic cells

    J. Immunol.

    (2004)
  • M.J. Androlewicz et al.

    Evidence that transporters associated with antigen processing translocate a major histocompatibility complex class I-binding peptide into the endoplasmic reticulum in an ATP-dependent manner

    Proc. Natl. Acad. Sci. USA

    (1993)
  • S.H. Baumeister et al.

    Coinhibitory pathways in immunotherapy for cancer

    Annu. Rev. Immunol.

    (2016)
  • W. Böhm et al.

    T cell-mediated, IFN-gamma-facilitated rejection of murine B16 melanomas

    J. Immunol.

    (1998)
  • W. Bracamonte-Baran et al.

    Modification of host dendritic cells by microchimerism-derived extracellular vesicles generates split tolerance

    Proc. Natl. Acad. Sci. USA

    (2017)
  • J. Canton et al.

    The receptor DNGR-1 signals for phagosomal rupture to promote cross-presentation of dead-cell-associated antigens

    Nat. Immunol.

    (2021)
  • P.J. Chefalo et al.

    Processing of exogenous antigens for presentation by Class I MHC molecules involves post-Golgi peptide exchange influenced by peptide-MHC complex stability and acidic pH

    J. Immunol.

    (2001)
  • L. Delamarre et al.

    Differential lysosomal proteolysis in antigen-presenting cells determines antigen fate

    Science

    (2005)
  • B.P. Dolan et al.

    Dendritic cells cross-dressed with peptide MHC Class I complexes prime CD8 + T cells

    J. Immunol.

    (2006)
  • M. Dougan et al.

    Cancer immunotherapy: Beyond checkpoint blockade

    Annu. Rev. Cancer Biol.

    (2019)
  • D. Dudziak et al.

    Differential antigen processing by dendritic cell subsets in vivo

    Science

    (2007)
  • E. Duong et al.

    Type I interferon activates MHC class I-dressed CD11b+ conventional dendritic cells to promote protective anti-tumor CD8+ T cell immunity

    Immunity

    (2021)
  • B.T. Edelson et al.

    Peripheral CD103+ dendritic cells form a unified subset developmentally related to CD8alpha+ conventional dendritic cells

    J. Exp. Med.

    (2010)
  • M.B. Fuertes et al.

    Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8{alpha}+ dendritic cells

    J. Exp. Med.

    (2011)
  • Z. Hein et al.

    Peptide-independent stabilization of MHC class I molecules breaches cellular quality control

    J. Cell Sci.

    (2014)
  • Cited by (47)

    • B7-H7: A potential target for cancer immunotherapy

      2023, International Immunopharmacology
    View all citing articles on Scopus
    7

    Lead contact

    View full text